Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 65: 1340-1351, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24120970

RESUMO

The antioxidant enzyme methionine sulfoxide reductase A (MsrA) is highly expressed in the retinal pigment epithelium (RPE), a support tissue for neighboring photoreceptors. MsrA protein levels correlate with sensitivity of RPE in culture to experimental oxidative stress. To investigate whether and how MsrA affects RPE functionality regardless of oxidative stress, we tested the effects of acute silencing or overexpression of MsrA on the phagocytosis of photoreceptor outer segment fragments (POS), a demanding, daily function of the RPE that is essential for vision. Endogenous MsrA localized to mitochondria and cytosol of rat RPE in culture. RPE cells manipulated to express higher or lower levels of MsrA than control cells showed no signs of cell death but increased or decreased, respectively, POS binding as well as engulfment. These effects of altered MsrA protein concentration on phagocytosis were independent of the levels of oxidative stress. However, altering MsrA expression had no effect on phagocytosis when mitochondrial respiration was inhibited. Furthermore, ATP content directly correlated with MsrA protein levels in RPE cells that used mitochondrial oxidative phosphorylation for ATP synthesis but not in RPE cells that relied on glycolysis alone. Overexpressing MsrA was sufficient to increase specifically the activity of complex IV of the respiratory chain, whereas activity of complex II and mitochondrial content were unaffected. Thus, MsrA probably enhances ATP synthesis by increasing complex IV activity. Such contribution of MsrA to energy metabolism is independent of its function in protection from elevated oxidative stress but contributes to routine but vital photoreceptor support by RPE cells.


Assuntos
Trifosfato de Adenosina/biossíntese , Mitocôndrias/metabolismo , Oxirredutases/imunologia , Fagocitose/imunologia , Epitélio Pigmentado da Retina/metabolismo , Animais , Antioxidantes , Linhagem Celular , Complexo II de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Metabolismo Energético/genética , Metabolismo Energético/imunologia , Células Epiteliais/citologia , Glicólise , Fosforilação Oxidativa , Estresse Oxidativo/imunologia , Oxirredutases/biossíntese , Oxirredutases/genética , Fagocitose/genética , Ligação Proteica/imunologia , Interferência de RNA , RNA Interferente Pequeno , Ratos , Segmento Externo das Células Fotorreceptoras da Retina/imunologia , Epitélio Pigmentado da Retina/citologia
2.
Drug Metab Dispos ; 39(6): 1014-21, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21383205

RESUMO

Sulindac is a nonsteroidal, anti-inflammatory drug (NSAID) that has also been studied for its anticancer activity. Recent studies suggest that sulindac and its metabolites act by sensitizing cancer cells to oxidizing agents and drugs that affect mitochondrial function, resulting in the production of reactive oxygen species and death by apoptosis. In contrast, normal cells are not killed under these conditions and, in some instances, are protected against oxidative stress. Sulindac has a methyl sulfoxide moiety with a chiral center and was used in all of the previous studies as a mixture of the R- and S-epimers. Because epimers of a compound can have very different chemical and biological properties, we have separated the R- and S-epimers of sulindac, studied their individual metabolism, and performed preliminary experiments on their effect on normal and lung cancer cells exposed to oxidative stress. Previous results had indicated that the reduction of (S)-sulindac to sulindac sulfide, the active NSAID, was catalyzed by methionine sulfoxide reductase (Msr) A. In the present study, we purified an enzyme that reduces (R)-sulindac and resembles MsrB in its substrate specificity. The oxidation of both epimers to sulindac sulfone is catalyzed primarily by the microsomal cytochrome P450 (P450) system, and the individual enzymes responsible have been identified. (S)-Sulindac increases the activity of the P450 system better than (R)-sulindac, but both epimers increase primarily the enzymes that oxidize (R)-sulindac. Both epimers can protect normal lung cells against oxidative damage and enhance the killing of lung cancer cells exposed to oxidative stress.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Metionina Sulfóxido Redutases/metabolismo , Sulindaco/metabolismo , Sulindaco/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Western Blotting , Cromatografia Líquida de Alta Pressão , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/genética , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Células Hep G2 , Humanos , Metionina Sulfóxido Redutases/química , Metionina Sulfóxido Redutases/genética , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Estereoisomerismo , Sulindaco/química , Sulindaco/farmacocinética
3.
J Clin Invest ; 121(3): 1174-90, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21285513

RESUMO

Acute lung injury (ALI) is a life-threatening condition in critically ill patients. Injury to the alveolar epithelium is a critical event in ALI, and accumulating evidence suggests that it is linked to proapoptotic Fas/FasL signals. Active soluble FasL (sFasL) is detectable in the bronchoalveolar lavage (BAL) fluid of patients with ALI, but the mechanisms controlling its bioactivity are unclear. We therefore investigated how the structure of sFasL influences cellular activation in human and mouse lungs and the role of oxidants and proteases in modifying sFasL activity. The sFasL in BAL fluid from patients with ALI was bioactive and present in high molecular weight multimers and aggregates. Oxidants generated from neutrophil myeloperoxidase in BAL fluid promoted aggregation of sFasL in vitro and in vivo. Oxidation increased the biological activity of sFasL at low concentrations but degraded sFasL at high concentrations. The amino-terminal extracellular stalk region of human sFasL was required to induce lung injury in mice, and proteolytic cleavage of the stalk region by MMP-7 reduced the bioactivity of sFasL in human cells in vitro. The sFasL recovered from the lungs of patients with ALI contained both oxidized methionine residues and the stalk region. These data provide what we believe to be new insights into the structural determinants of sFasL bioactivity in the lungs of patients with ALI.


Assuntos
Proteína Ligante Fas/metabolismo , Pulmão/metabolismo , Animais , Apoptose , Lavagem Broncoalveolar , Líquido da Lavagem Broncoalveolar , Dano ao DNA , Humanos , Peróxido de Hidrogênio/química , Metaloproteinase 7 da Matriz/metabolismo , Metionina/metabolismo , Camundongos , Oxidantes/metabolismo , Oxigênio/metabolismo , Estrutura Terciária de Proteína
4.
Free Radic Res ; 42(11-12): 978-88, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19085252

RESUMO

Methionine sulphoxide reductase A (MSRA) that reduces methionine-S-sulphoxide back to methionine constitutes a catalytic antioxidant mechanism to prevent oxidative damage at multiple sub-cellular loci. This study examined the relative importance of protection of the cytoplasm and mitochondria by MSRA using A-10 vascular smooth muscle cells, a cell type that requires a low level of reactive oxygen species (ROS) for normal function but is readily damaged by higher concentrations of ROS. Adenoviral over-expression of human MSRA variants, targeted to either mitochondria or the cytoplasm, did not change basal viability of non-stressed cells. Oxidative stress caused by treatment with the methionine-preferring oxidizing reagent chloramine-T decreased cell viability in a concentration-dependent manner. Cytoplasmic MSRA preserved cell viability more effectively than mitochondrial MSRA and co-application of S-methyl-L-cysteine, an amino acid that acts as a substrate for MSRA when oxidized, further increased the extent of protection. This suggests an important role for an MSRA catalytic antioxidant cycle for protection of the cytoplasmic compartment against oxidative damage.


Assuntos
Cisteína/farmacologia , Regulação Enzimológica da Expressão Gênica , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Oxirredutases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cloraminas/farmacologia , Cisteína/análogos & derivados , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Metionina Sulfóxido Redutases , Estresse Oxidativo , Oxirredutases/genética , Ratos , Compostos de Tosil/farmacologia
5.
J Neurosci ; 27(47): 12808-16, 2007 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18032652

RESUMO

Parkinson's disease (PD), a common neurodegenerative disease, is caused by loss of dopaminergic neurons in the substantia nigra. Although the underlying cause of the neuronal loss is unknown, oxidative stress is thought to play a major role in the pathogenesis of PD. The amino acid methionine is readily oxidized to methionine sulfoxide, and its reduction is catalyzed by a family of enzymes called methionine sulfoxide reductases (MSRs). The reversible oxidation-reduction cycle of methionine involving MSRs has been postulated to act as a catalytic antioxidant system protecting cells from oxidative damage. Here, we show that one member of the MSR family, MSRA, inhibits development of the locomotor and circadian rhythm defects caused by ectopic expression of human alpha-synuclein in the Drosophila nervous system. Furthermore, we demonstrate that one way to enhance the MSRA antioxidant system is dietary supplementation with S-methyl-L-cysteine (SMLC), found abundantly in garlic, cabbage, and turnips. SMLC, a substrate in the catalytic antioxidant system mediated by MSRA, prevents the alpha-synuclein-induced abnormalities. Therefore, interventions focusing on the enzymatic reduction of oxidized methionine catalyzed by MSRA represent a new prevention and therapeutic approach for PD and potentially for other neurodegenerative diseases involving oxidative stress.


Assuntos
Cisteína/análogos & derivados , Suplementos Nutricionais , Oxirredutases/administração & dosagem , Doença de Parkinson/prevenção & controle , Animais , Cisteína/administração & dosagem , Drosophila , Humanos , Metionina Sulfóxido Redutases , Atividade Motora/fisiologia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo
6.
Proc Natl Acad Sci U S A ; 104(23): 9597-602, 2007 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-17535911

RESUMO

The reduction of methionine sulfoxide (MetO) is mediated by methionine sulfoxide reductases (Msr). The MsrA and MsrB families can reduce free MetO and MetO within a peptide or protein context. This process is stereospecific with the S- and R-forms of MetO repaired by MsrA and MsrB, respectively. Cell extracts from an MsrA(-)B(-) knockout of Escherichia coli have several remaining Msr activities. This study has identified an enzyme specific for the free form of Met-(R)-O, fRMsr, through proteomic analysis. The recombinant enzyme exhibits the same substrate specificity and is as active as MsrA family members. E. coli fRMsr is, however, 100- to 1,000-fold more active than non-selenocysteine-containing MsrB enzymes for free Met-(R)-O. The crystal structure of E. coli fRMsr was previously determined, but no known function was assigned. Thus, the function of this protein has now been determined. The structural similarity of the E. coli and yeast proteins suggests that most fRMsrs use three cysteine residues for catalysis and the formation of a disulfide bond to enclose a small active site cavity. This latter feature is most likely a key determinant of substrate specificity. Moreover, E. coli fRMsr is the first GAF domain family member to show enzymatic activity. Other GAF domain proteins substitute the Cys residues and others to specifically bind cyclic nucleotides, chromophores, and many other ligands for signal potentiation. Therefore, Met-(R)-O may represent a signaling molecule in response to oxidative stress and nutrients via the TOR pathway in some organisms.


Assuntos
Escherichia coli/enzimologia , Modelos Moleculares , Oxirredutases/metabolismo , Proteínas Recombinantes/metabolismo , Sequência de Aminoácidos , Biologia Computacional , Cinética , Espectrometria de Massas , Metionina/análogos & derivados , Metionina/metabolismo , Metionina Sulfóxido Redutases , Dados de Sequência Molecular , Oxirredutases/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Análise de Sequência de DNA , Especificidade por Substrato
7.
J Biol Chem ; 281(43): 32668-75, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-16926157

RESUMO

The PilB protein from Neisseria gonorrhoeae is located in the periplasm and made up of three domains. The N-terminal, thioredoxin-like domain (NT domain) is fused to tandem methionine sulfoxide reductase A and B domains (MsrA/B). We show that the alpha domain of Escherichia coli DsbD is able to reduce the oxidized NT domain, which suggests that DsbD in Neisseria can transfer electrons from the cytoplasmic thioredoxin to the periplasm for the reduction of the MsrA/B domains. An analysis of the available complete genomes provides further evidence for this proposition in other bacteria where DsbD/CcdA, Trx, MsrA, and MsrB gene homologs are all located in a gene cluster with a common transcriptional direction. An examination of wild-type PilB and a panel of Cys to Ser mutants of the full-length protein and the individually expressed domains have also shown that the NT domain more efficiently reduces the MsrA/B domains when in the polyprotein context. Within this frame-work there does not appear to be a preference for the NT domain to reduce the proximal MsrA domain over MsrB domain. Finally, we report the 1.6A crystal structure of the NT domain. This structure confirms the presence of a surface loop that makes it different from other membrane-tethered, Trx-like molecules, including TlpA, CcmG, and ResA. Subtle differences are observed in this loop when compared with the Neisseria meningitidis NT domain structure. The data taken together supports the formation of specific NT domain interactions with the MsrA/B domains and its in vivo recycling partner, DsbD.


Assuntos
Elétrons , Proteínas de Escherichia coli/química , Neisseria gonorrhoeae/química , Oxirredutases/química , Oxirredutases/metabolismo , Tiorredoxinas/química , Sítios de Ligação , Cristalografia por Raios X , Cisteína/química , Metionina Sulfóxido Redutases , Modelos Químicos , Modelos Moleculares , Oxirredução , Estrutura Terciária de Proteína , Tiorredoxinas/isolamento & purificação
8.
Proc Natl Acad Sci U S A ; 103(23): 8656-61, 2006 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-16735467

RESUMO

It has been generally accepted, primarily from studies on methionine sulfoxide reductase (Msr) A, that the biological reducing agent for the members of the Msr family is reduced thioredoxin (Trx), although high levels of DTT can be used as the reductant in vitro. Preliminary experiments using both human recombinant MsrB2 (hMsrB2) and MsrB3 (hMsrB3) showed that although DTT can function in vitro as the reducing agent, Trx works very poorly, prompting a more careful comparison of the ability of DTT and Trx to function as reducing agents with the various members of the Msr family. Escherichia coli MsrA and MsrB and bovine MsrA efficiently use either Trx or DTT as reducing agents. In contrast, hMsrB2 and hMsrB3 show <10% of the activity with Trx as compared with DTT, raising the possibility that, in animal cells, Trx may not be the direct hydrogen donor or that there may be a Trx-independent reducing system required for MsrB2 and MsrB3 activity. A heat-stable protein has been detected in bovine liver that, in the presence of EDTA, can support the Msr reaction in the absence of either Trx or DTT. This protein has been identified as a zinc-containing metallothionein (Zn-MT). The results indicate that thionein (T), which is formed when the zinc is removed from Zn-MT, can function as a reducing system for the Msr proteins because of its high content of cysteine residues and that Trx can reduce oxidized T.


Assuntos
Ergotioneína/metabolismo , Oxirredutases/metabolismo , Animais , Bovinos , Ditiotreitol/farmacologia , Ácido Edético/farmacologia , Ergotioneína/farmacologia , Humanos , Fígado/enzimologia , Metalotioneína/metabolismo , Metionina Sulfóxido Redutases , Oxirredução/efeitos dos fármacos , Oxirredutases/isolamento & purificação , Ratos , Proteínas Recombinantes/metabolismo , Tiorredoxinas/metabolismo , Zinco/metabolismo
9.
Invest Ophthalmol Vis Sci ; 46(6): 2107-12, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15914630

RESUMO

PURPOSE: Methionine-sulfoxide reductases are unique, in that their ability to repair oxidized proteins and MsrA, which reduces S-methionine sulfoxide, can protect lens cells against oxidative stress damage. To date, the roles of MsrB1, -B2 and -B3 which reduce R-methionine sulfoxide have not been established for any mammalian system. The present study was undertaken to identify those MsrBs expressed by the lens and to evaluate the enzyme activities, expression patterns, and abilities of the identified genes to defend lens cells against oxidative stress damage. METHODS: Enzyme activities were determined with bovine lens extracts. The identities and spatial expression patterns of MsrB1, -B2, and -B3 transcripts were examined by RT-PCR in human lens and 21 other tissues. Oxidative stress resistance was measured using short interfering (si)RNA-mediated gene-silencing in conjunction with exposure to tert-butyl hydroperoxide (tBHP) and MTS viability measurements in SRA04/01 human lens epithelial cells. RESULTS: Forty percent of the Msr enzyme activity present in the lens was MsrB, whereas the remaining enzyme activity was MsrA. MsrB1 (selenoprotein R, localized in the cytosol and nucleus), MsrB2 (CBS-1, localized in the mitochondria), and MsrB3 (localized in the endoplasmic reticulum and mitochondria) were all expressed by the lens. These genes exhibit asymmetric expression patterns between different human tissues and different lens sublocations, including lens fibers. All three genes are required for lens cell viability, and their silencing in lens cells results in increased oxidative-stress-induced cell death. CONCLUSIONS: The present data suggest important roles for both MsrA and -Bs in lens cell viability and oxidative stress protection. The differential tissue distribution and lens expression patterns of these genes, coupled with increased oxidative-stress-induced cell death on their deletion provides evidence that they are important for lens cell function, resistance to oxidative stress, and, potentially, cataractogenesis.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Cristalino/enzimologia , Estresse Oxidativo , Oxirredutases/genética , Oxirredutases/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Sobrevivência Celular , Células Cultivadas , Células Epiteliais/enzimologia , Inativação Gênica/fisiologia , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Cristalino/citologia , Metionina Sulfóxido Redutases , Proteínas dos Microfilamentos , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual , Fatores de Transcrição , terc-Butil Hidroperóxido/farmacologia
10.
Proc Natl Acad Sci U S A ; 101(5): 1159-64, 2004 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-14745014

RESUMO

Hypoxia/reoxygenation induces cellular injury by promoting oxidative stress. Reversible oxidation of methionine in proteins involving the enzyme peptide methionine sulfoxide reductase type A (MSRA) is postulated to serve a general antioxidant role. Therefore, we examined whether overexpression of MSRA protected cells from hypoxia/reoxygenation injury. Brief hypoxia increased the intracellular reactive oxygen species (ROS) level in PC12 cells and promoted apoptotic cell death. Adenovirus-mediated overexpression of MSRA significantly diminished the hypoxia-induced increase in ROS and facilitated cell survival. Measurements of the membrane potentials of intact mitochondria in PC12 cells and of isolated rat liver mitochondria showed that hypoxia induced depolarization of the mitochondrial membrane. The results demonstrate that MSRA plays a protective role against hypoxia/reoxygenation-induced cell injury and suggest the therapeutic potential of MSRA in ischemic heart and brain disease.


Assuntos
Hipóxia Celular , Citoproteção , Metionina/análogos & derivados , Neurônios/metabolismo , Oxirredutases/fisiologia , Animais , Apoptose , Potenciais da Membrana , Metionina/análise , Metionina Sulfóxido Redutases , Mitocôndrias/fisiologia , Neurônios/patologia , Células PC12 , Ratos , Espécies Reativas de Oxigênio
11.
Biochem Biophys Res Commun ; 312(4): 1005-10, 2003 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-14651971

RESUMO

Sulindac is a known anti-inflammatory drug that functions by inhibition of cyclooxygenases 1 and 2 (COX). There has been recent interest in Sulindac and other non-steroidal anti-inflammatory drugs (NSAID) because of their anti-tumor activity against colorectal cancer. Studies with sulindac have indicated that it may also function as an anti-tumor agent by stimulating apoptosis. Sulindac is a pro-drug, containing a methyl sulfoxide group, that must be reduced to sulindac sulfide to be active as a COX inhibitor. In the present studies we have developed a simple assay to measure sulindac reduction and tested sulindac as a substrate for 6 known members of the methionine sulfoxide reductase (Msr) family that have been identified in Escherichia coli. Only MsrA and a membrane associated Msr can reduce sulindac to the active sulfide. The reduction of sulindac also has been demonstrated in extracts of calf liver, kidney, and brain. Sulindac reductase activity is also present in mitochondria and microsomes.


Assuntos
Encéfalo/metabolismo , Escherichia coli/química , Rim/metabolismo , Fígado/metabolismo , Oxirredutases/química , Oxirredutases/metabolismo , Sulindaco/análogos & derivados , Sulindaco/química , Sulindaco/metabolismo , Animais , Anti-Inflamatórios não Esteroides/metabolismo , Bovinos , Cromatografia/métodos , Ativação Enzimática , Escherichia coli/enzimologia , Metionina Sulfóxido Redutases , Microssomos/metabolismo , Mitocôndrias/metabolismo , Especificidade de Órgãos , Oxirredução , Oxirredutases/classificação , Sulindaco/análise
12.
Ann N Y Acad Sci ; 987: 68-78, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12727625

RESUMO

Genetic studies in mice indicate that predisposition to lupus-like diseases is caused by at least three mechanisms: (1) alterations in the threshold of activation of lymphocytes or macrophages; (2) defective signaling for activation-induced cell death; and (3) reduced clearance of apoptotic cells. To define the mechanisms whereby lupus develops in mice with deficiencies in either C1q, serum amyloid P component (SAP, the mouse counterpart of C-reactive protein, or CRP), or serum IgM, we studied the efficiency of phagocytosis of apoptotic cells using serum with varying levels of C1q, CRP, or IgM; we also examined the immune response to ingestion of dying cells under these conditions. Deficiency of C1q led to impaired macrophage phagocytosis of apoptotic cells, whereas CRP augmented phagocytosis, largely through recruitment of the early complement components. Like CRP, normal polyclonal IgM bound to apoptotic cells and activated complement on the cell surface. Similarly, direct binding as well as absorption experiments revealed that CRP and IgM antibodies had a similar ligand recognition specificity, namely lysophospholipids containing phosphorylcholine. IL-12 provides a pivotal link between macrophages and the T cell response to ingested material. We observed that necrotic cells induced IL-12 p40 expression, whereas phagocytosis of apoptotic cells profoundly reduced IL-12 production from stimulated macrophages. Furthermore, soluble factors from macrophages that had ingested apoptotic cells suppressed interferon-gamma production by activated T cells. These findings suggest that phospholipid exposure on apoptotic cells promotes opsonization by serum proteins leading to activation of complement, macrophage ingestion, and T cell suppression. We discuss how deficient opsonization or processing of dying cells leads to autoimmunity.


Assuntos
Apoptose/imunologia , Macrófagos/imunologia , Proteínas Opsonizantes/imunologia , Linfócitos T/imunologia , Autoanticorpos/imunologia , Proteínas do Sistema Complemento/imunologia , Imunoglobulina M/imunologia
13.
Biochem Biophys Res Commun ; 302(2): 284-9, 2003 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-12604343

RESUMO

It is known that reactive oxygen species can oxidize methionine residues in proteins in a non-stereospecific manner, and cells have mechanisms to reverse this damage. MsrA and MsrB are members of the methionine sulfoxide family of enzymes that specifically reduce the S and R forms, respectively, of methionine sulfoxide in proteins. However, in Escherichia coli the level of MsrB activity is very low which suggested that there may be other enzymes capable of reducing the R epimer of methionine sulfoxide in proteins. Employing a msrA/B double mutant, a new peptide methionine sulfoxide reductase activity has been found associated with membrane vesicles from E. coli. Both the R and S forms of N-acetylmethionine sulfoxide, D-ala-met(o)-enkephalin and methionine sulfoxide, are reduced by this membrane associated activity. The reaction requires NADPH and may explain, in part, how the R form of methionine sulfoxide in proteins is reduced in E. coli. In addition, a new soluble Msr activity was also detected in the soluble extracts of the double mutant that specifically reduces the S epimer of met(o) in proteins.


Assuntos
Escherichia coli/enzimologia , Oxirredutases/metabolismo , Membrana Celular/enzimologia , Metionina Sulfóxido Redutases , Oxirredução , Oxirredutases/classificação , Oxirredutases/genética , Peptídeos/metabolismo , Solubilidade
14.
Proc Natl Acad Sci U S A ; 99(15): 10108-13, 2002 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-12096194

RESUMO

The PilB protein of Neisseria gonorrhoeae has been reported to be involved in the regulation of pilin gene transcription, but it also possesses significant homology to the peptide methionine sulfoxide reductase family of enzymes, specifically MsrA and MsrB from Escherichia coli. MsrA and MsrB in E. coli are able to reduce methionine sulfoxide residues in proteins to methionines. In addition, the gonococcal PilB protein encodes for both MsrA and MsrB activity associated with the repair of oxidative damage to proteins. In this work, we demonstrate that the PilB protein of Neisseria gonorrhoeae is not involved in pilus expression. Additionally, we show that wild-type N. gonorrhoeae produces two forms of this polypeptide, one of which contains a signal sequence and is secreted from the bacterial cytoplasm to the outer membrane; the other lacks a signal sequence and is cytoplasmic. Furthermore, we show that the secreted form of the PilB protein is involved in survival in the presence of oxidative damage.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas de Membrana Transportadoras , Neisseria gonorrhoeae/fisiologia , Oxirredutases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Fosfatase Alcalina/análise , Proteínas de Bactérias/genética , Primers do DNA , Fímbrias Bacterianas/fisiologia , Fímbrias Bacterianas/ultraestrutura , Peróxido de Hidrogênio/farmacologia , Metionina Sulfóxido Redutases , Mutagênese , Mutagênese Insercional , Neisseria gonorrhoeae/citologia , Neisseria gonorrhoeae/efeitos dos fármacos , Neisseria gonorrhoeae/genética , Oxirredutases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Nat Struct Biol ; 9(5): 348-52, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11938352

RESUMO

Methionine sulfoxide reductases (Msr) protect against oxidative damage that can contribute to cell death. The tandem Msr domains (MsrA and MsrB) of the pilB protein from Neisseria gonorrhoeae each reduce different epimeric forms of methionine sulfoxide. The overall fold of the MsrB domain revealed by the 1.85 A crystal structure shows no resemblance to the previously determined MsrA structures from other organisms. Despite the lack of homology, the active sites show approximate mirror symmetry. In each case, conserved amino acid motifs mediate the stereo-specific recognition and reduction of the substrate. Unlike the MsrA domain, the MsrB domain activates the cysteine or selenocysteine nucleophile through a unique Cys-Arg-Asp/Glu catalytic triad. The collapse of the reaction intermediate most likely results in the formation of a sulfenic or selenenic acid moiety. Regeneration of the active site occurs through a series of thiol-disulfide exchange steps involving another active site Cys residue and thioredoxin. These observations have broad implications for modular catalysis, antibiotic drug design and continuing longevity studies in mammals.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Neisseria gonorrhoeae/enzimologia , Oxirredutases/química , Oxirredutases/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Metionina Sulfóxido Redutases , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Alinhamento de Sequência , Relação Estrutura-Atividade
16.
Proc Natl Acad Sci U S A ; 99(5): 2748-53, 2002 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-11867705

RESUMO

Cumulative oxidative damages to cell constituents are considered to contribute to aging and age-related diseases. The enzyme peptide methionine sulfoxide reductase A (MSRA) catalyzes the repair of oxidized methionine in proteins by reducing methionine sulfoxide back to methionine. However, whether MSRA plays a role in the aging process is poorly understood. Here we report that overexpression of the msrA gene predominantly in the nervous system markedly extends the lifespan of the fruit fly Drosophila. The MSRA transgenic animals are more resistant to paraquat-induced oxidative stress, and the onset of senescence-induced decline in the general activity level and reproductive capacity is delayed markedly. The results suggest that oxidative damage is an important determinant of lifespan, and MSRA may be important in increasing the lifespan in other organisms including humans.


Assuntos
Longevidade , Oxirredutases/fisiologia , Animais , Animais Geneticamente Modificados , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Longevidade/fisiologia , Metionina Sulfóxido Redutases , Oxirredutases/genética , Peptídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA